Jiang Xiaotao, Wu Anzhou, Yan Jiaxing, Chen Jingming, Wen Yi, Wu Hui, Yan Ning, Yang Zehong, Liu Fengbin, Li Peiwu
First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China.
First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China; The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China.
Int Immunopharmacol. 2025 Jun 26;159:114907. doi: 10.1016/j.intimp.2025.114907. Epub 2025 May 22.
An immune-suppressive tumor microenvironment (TME) that encourages tumor growth is a hallmark of gastric cancer (GC), which is implicated in the development, metastasis, and unfavorable prognosis of GC. Acanthopanax senticosus (Rupr.&Maxim.) Harms (AS), also called Siberian Ginseng (Chinese: Ci wu jia), is a commonly used traditional Chinese herbal medicine with immune-enhancing, anti-tumor, anti-fatigue, neuroregulatory, blood circulation-improving, and antioxidant properties. Recently, it has also been demonstrated to improve anti-tumor immunity in GC. Eleutheroside A (EA), one of the primary bioactive saponins of AS, has immunoregulatory functions. Given the immunomodulatory and anti-tumor effects of EA, it is crucial to investigate its regulatory impact on the immune landscape of GC.
To determine the effects of EA on immune responses in GC, a subcutaneous GC mouse model was established. Tumor growth, body weight changes, and immune responses in the mice treated with EA were measured. The proportion of CD4T, CD8T, B cells, NK cells, TAMs, DCs and MDSCs in the spleens were analyzed using flow cytometry. MDSCs and CD4/CD8 T cell infiltration in tumor tissue were analyzed using immunofluorescence. Bulk RNA sequencing (bulk RNA-seq) data from the Cancer Genome Atlas (TCGA) and two single-cell RNA sequencing (scRNA-seq) datasets (accession numbers GSE183904 and GSE150290) were used to examine changes in MDSCs and T cell infiltration within the TME of GC and to identify MDSCs-related targets. Network pharmacology analysis, protein-protein interaction (PPI) network analysis, dynamics simulations, molecular docking and surface plasmon resonance (SPR) were applied to explore the potential mechanisms underlying EA's intervention in MDSCs. Flow cytometry, qPCR, and western blotting and Seahorse assays were applied for analyzing MDSCs isolated from in vivo and in vitro-induced conditions, aiming to delineate the mechanism of EA on MDSCs glycolysis and immunosuppressive functions mediated by the PI3K/AKT1/mTOR signaling pathway.
In vivo, EA treatment effectively suppressed GC tumor growth and progression in mice, reducing the prevalence of MDSCs and increasing CD4/CD8 T cell levels. In vitro, EA not only decreased the frequency of MDSCs but also alleviated their immune-suppressing capabilities on CD4/CD8 T cells. Network pharmacology, coupled with scRNA-seq analysis, dynamic simulations, and molecular docking studies, suggested that EA might modulate the PI3K/AKT1/mTOR signaling pathway to influence glycolysis in MDSCs. Surface plasmon resonance (SPR) analysis confirmed that EA directly interacts with AKT1. Further validation experiments revealed that in the GC TME, EA treatment decreased the expression of p-PI3K, p-AKT1, p-mTOR, HIF1α, as well as glycolytic genes and glycolytic activity in MDSCs. Additionally, EA led to the downregulation of p-STAT3 and its downstream immunosuppressive factors within these cells. Restoring AKT1 activation could reverse the inhibitory effects of EA on MDSCs glycolysis and the downregulation of immunosuppressive molecules. Moreover, HIF-1α inhibition abolished EA's inhibitory effects on MDSCs.
EA can attenuate the immune-suppressive capacity of MDSCs in GC by inhibiting the PI3K/AKT1/mTOR pathway and suppressing HIF-1α-mediated glycolysis, thereby offering a novel therapeutic approach to targeting the immune-suppressive microenvironment in GC.
促进肿瘤生长的免疫抑制性肿瘤微环境(TME)是胃癌(GC)的一个标志,它与GC的发生、转移及不良预后有关。刺五加(Acanthopanax senticosus (Rupr.&Maxim.) Harms,AS),也叫西伯利亚人参(中文:刺五加),是一种常用的传统中草药,具有免疫增强、抗肿瘤、抗疲劳、神经调节、改善血液循环和抗氧化特性。最近,也已证明它可改善GC中的抗肿瘤免疫。刺五加苷A(EA)是AS的主要生物活性皂苷之一,具有免疫调节功能。鉴于EA的免疫调节和抗肿瘤作用,研究其对GC免疫格局的调节影响至关重要。
为确定EA对GC免疫反应的影响,建立了皮下GC小鼠模型。测量了用EA处理的小鼠的肿瘤生长、体重变化和免疫反应。使用流式细胞术分析脾脏中CD4T、CD8T、B细胞、NK细胞、肿瘤相关巨噬细胞(TAMs)、树突状细胞(DCs)和髓系来源的抑制性细胞(MDSCs)的比例。使用免疫荧光分析肿瘤组织中MDSCs和CD4/CD8 T细胞浸润情况。来自癌症基因组图谱(TCGA)的批量RNA测序(bulk RNA-seq)数据以及两个单细胞RNA测序(scRNA-seq)数据集(登录号GSE183904和GSE150290)用于检查GC的TME内MDSCs和T细胞浸润的变化,并鉴定与MDSCs相关的靶点。应用网络药理学分析、蛋白质-蛋白质相互作用(PPI)网络分析、动力学模拟、分子对接和表面等离子体共振(SPR)来探索EA干预MDSCs的潜在机制。应用流式细胞术、qPCR、蛋白质免疫印迹法和海马实验分析从体内和体外诱导条件下分离的MDSCs,旨在阐明EA对由PI3K/AKT1/mTOR信号通路介导的MDSCs糖酵解和免疫抑制功能的作用机制。
在体内,EA治疗有效抑制了小鼠GC肿瘤的生长和进展,降低了MDSCs的比例并提高了CD4/CD8 T细胞水平。在体外,EA不仅降低了MDSCs的频率,还减轻了它们对CD4/CD8 T细胞的免疫抑制能力。网络药理学结合scRNA-seq分析、动力学模拟和分子对接研究表明,EA可能通过调节PI3K/AKT1/mTOR信号通路来影响MDSCs中的糖酵解。表面等离子体共振(SPR)分析证实EA直接与AKT1相互作用。进一步的验证实验表明,在GC的TME中,EA处理降低了p-PI3K、p-AKT1、p-mTOR、HIF1α的表达,以及MDSCs中的糖酵解基因和糖酵解活性。此外,EA导致这些细胞中p-STAT3及其下游免疫抑制因子的下调。恢复AKT1的激活可以逆转EA对MDSCs糖酵解的抑制作用以及免疫抑制分子的下调。此外,HIF-1α的抑制消除了EA对MDSCs的抑制作用。
EA可通过抑制PI3K/AKT1/mTOR途径并抑制HIF-1α介导的糖酵解来减弱GC中MDSCs的免疫抑制能力,从而为靶向GC免疫抑制微环境提供一种新的治疗方法。